51
|
Gao J, Wang Y, Cai M, Pan Y, Xu H, Jiang J, Ji H, Wang H. Mechanistic insights into EGFR membrane clustering revealed by super-resolution imaging. NANOSCALE 2015; 7:2511-9. [PMID: 25569174 DOI: 10.1039/c4nr04962d] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The clustering of membrane receptors such as EGFR is critical for various biological processes, for example cell signaling and tumorigenesis. However, the mechanism involved remains poorly understood. Here, we used a super resolution imaging technique, which has shattered the longstanding resolution barrier of light diffraction, to investigate the distribution of membrane EGFR on apical or basal surfaces of COS-7 cells and on the surface of suspended COS-7 cells. Our data show that more and larger EGFR clusters are detected on the apical surface in comparison with those on the basal surface and this difference is not affected by the EGFR activation state, whereas suspended COS-7 cells exhibit a moderate clustering state and a homogeneous distribution pattern, indicating that the external environment surrounding the cell membrane is the decisive factor in the EGFR clustering pattern. A dual-color dSTORM image reveals the significant colocalization of EGFR and lipid rafts; interestingly MβCD treatment leads to a dramatic decrease of the amount and size of EGFR clusters on both apical and basal surfaces, highlighting a key role of lipid rafts in EGFR cluster formation. Altogether, our results illustrate the distribution pattern of EGFR in polarized cells and uncover the essential role of lipid rafts in EGFR cluster maintenance.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Tardif JC, Rhéaume E, Lemieux Perreault LP, Grégoire JC, Feroz Zada Y, Asselin G, Provost S, Barhdadi A, Rhainds D, L'Allier PL, Ibrahim R, Upmanyu R, Niesor EJ, Benghozi R, Suchankova G, Laghrissi-Thode F, Guertin MC, Olsson AG, Mongrain I, Schwartz GG, Dubé MP. Pharmacogenomic determinants of the cardiovascular effects of dalcetrapib. ACTA ACUST UNITED AC 2015; 8:372-82. [PMID: 25583994 DOI: 10.1161/circgenetics.114.000663] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/16/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Dalcetrapib did not improve clinical outcomes, despite increasing high-density lipoprotein cholesterol by 30%. These results differ from other evidence supporting high-density lipoprotein as a therapeutic target. Responses to dalcetrapib may vary according to patients' genetic profile. METHODS AND RESULTS We conducted a pharmacogenomic evaluation using a genome-wide approach in the dal-OUTCOMES study (discovery cohort, n=5749) and a targeted genotyping panel in the dal-PLAQUE-2 imaging trial (support cohort, n=386). The primary endpoint for the discovery cohort was a composite of cardiovascular events. The change from baseline in carotid intima-media thickness on ultrasonography at 6 and 12 months was evaluated as supporting evidence. A single-nucleotide polymorphism was found to be associated with cardiovascular events in the dalcetrapib arm, identifying the ADCY9 gene on chromosome 16 (rs1967309; P=2.41×10(-8)), with 8 polymorphisms providing P<10(-6) in this gene. Considering patients with genotype AA at rs1967309, there was a 39% reduction in the composite cardiovascular endpoint with dalcetrapib compared with placebo (hazard ratio, 0.61; 95% confidence interval, 0.41-0.92). In patients with genotype GG, there was a 27% increase in events with dalcetrapib versus placebo. Ten single-nucleotide polymorphism in the ADCY9 gene, the majority in linkage disequilibrium with rs1967309, were associated with the effect of dalcetrapib on intima-media thickness (P<0.05). Marker rs2238448 in ADCY9, in linkage disequilibrium with rs1967309 (r(2)=0.8), was associated with both the effects of dalcetrapib on intima-media thickness in dal-PLAQUE-2 (P=0.009) and events in dal-OUTCOMES (P=8.88×10(-8); hazard ratio, 0.67; 95% confidence interval, 0.58-0.78). CONCLUSIONS The effects of dalcetrapib on atherosclerotic outcomes are determined by correlated polymorphisms in the ADCY9 gene. CLINICAL TRIAL INFORMATION URL: http://www.clinicaltrials.gov. Unique identifiers: NCT00658515 and NCT01059682.
Collapse
Affiliation(s)
- Jean-Claude Tardif
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.).
| | - Eric Rhéaume
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Louis-Philippe Lemieux Perreault
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Jean C Grégoire
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Yassamin Feroz Zada
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Géraldine Asselin
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Sylvie Provost
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Amina Barhdadi
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - David Rhainds
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Philippe L L'Allier
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Reda Ibrahim
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Ruchi Upmanyu
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Eric J Niesor
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Renée Benghozi
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Gabriela Suchankova
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Fouzia Laghrissi-Thode
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Marie-Claude Guertin
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Anders G Olsson
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Ian Mongrain
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Gregory G Schwartz
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.)
| | - Marie-Pierre Dubé
- Montreal Heart Institute (J.-C.T., E.R., L.-P.L.P., J.C.G., Y.F.Z., G.A., S.P., A.B., D.R., P.L.L'., R.I., M.-C.G., I.M., M.-P.D.), Université de Montréal (J.-C.T., E.R., J.C.G., P.L.L'., R.I., M.-P.D.), Université de Montréal Beaulieu-Saucier Pharmacogenomics, Centre Montreal, Quebec, Canada (L.-P.L.P., Y.F.Z., G.A., S.P., A.B., I.M., M.-P.D.), Montreal Health Innovations Coordinating Centre (MHICC) (M.-C.G.), Montreal, Quebec, Canada; Stockholm Heart Center, Stockholm, Sweden (A.G.O.); Veterans Affairs Medical Center, University of Colorado, Denver (G.G.S.); and F. Hoffmann-La Roche, Basel, Switzerland (R.U., E.J.N., R.B., G.S., F.L.-T.).
| |
Collapse
|
53
|
Tsvetanova NG, von Zastrow M. Spatial encoding of cyclic AMP signaling specificity by GPCR endocytosis. Nat Chem Biol 2014; 10:1061-5. [PMID: 25362359 PMCID: PMC4232470 DOI: 10.1038/nchembio.1665] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022]
Abstract
G protein-coupled receptors (GPCRs) are well known to signal via cyclic AMP (cAMP) production at the plasma membrane, but it is now clear that various GPCRs also signal after internalization. Apart from its temporal impact through prolonging the cellular response, we wondered whether the endosome-initiated signal encodes any discrete spatial information. Using the β2-adrenoceptor (β2-AR) as a model, we show that endocytosis is required for the full repertoire of downstream cAMP-dependent transcriptional control. Next, we describe an orthogonal optogenetic approach to definitively establish that the location of cAMP production is indeed the critical variable determining the transcriptional response. Finally, our results suggest that this spatial encoding scheme helps cells functionally discriminate chemically distinct β2-AR ligands according to differences in their ability to promote receptor endocytosis. These findings reveal a discrete principle for achieving cellular signaling specificity based on endosome-mediated spatial encoding of intracellular second messenger production and 'location-aware' downstream transcriptional control.
Collapse
Affiliation(s)
| | - Mark von Zastrow
- Department of Psychiatry, University of California, San Francisco CA, USA
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco CA, USA
| |
Collapse
|
54
|
Prasanna X, Chattopadhyay A, Sengupta D. Cholesterol modulates the dimer interface of the β₂-adrenergic receptor via cholesterol occupancy sites. Biophys J 2014; 106:1290-300. [PMID: 24655504 DOI: 10.1016/j.bpj.2014.02.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 02/01/2014] [Accepted: 02/03/2014] [Indexed: 11/26/2022] Open
Abstract
The β2-adrenergic receptor is an important member of the G-protein-coupled receptor (GPCR) superfamily, whose stability and function are modulated by membrane cholesterol. The recent high-resolution crystal structure of the β2-adrenergic receptor revealed the presence of possible cholesterol-binding sites in the receptor. However, the functional relevance of cholesterol binding to the receptor remains unexplored. We used MARTINI coarse-grained molecular-dynamics simulations to explore dimerization of the β2-adrenergic receptor in lipid bilayers containing cholesterol. A novel (to our knowledge) aspect of our results is that receptor dimerization is modulated by membrane cholesterol. We show that cholesterol binds to transmembrane helix IV, and cholesterol occupancy at this site restricts its involvement at the dimer interface. With increasing cholesterol concentration, an increased presence of transmembrane helices I and II, but a reduced presence of transmembrane helix IV, is observed at the dimer interface. To our knowledge, this study is one of the first to explore the correlation between cholesterol occupancy and GPCR organization. Our results indicate that dimer plasticity is relevant not just as an organizational principle but also as a subtle regulatory principle for GPCR function. We believe these results constitute an important step toward designing better drugs for GPCR dimer targets.
Collapse
|
55
|
Tilley DG, Zhu W, Myers VD, Barr LA, Gao E, Li X, Song J, Carter RL, Makarewich CA, Yu D, Troupes CD, Grisanti LA, Coleman RC, Koch WJ, Houser SR, Cheung JY, Feldman AM. β-adrenergic receptor-mediated cardiac contractility is inhibited via vasopressin type 1A-receptor-dependent signaling. Circulation 2014; 130:1800-11. [PMID: 25205804 DOI: 10.1161/circulationaha.114.010434] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Enhanced arginine vasopressin levels are associated with increased mortality during end-stage human heart failure, and cardiac arginine vasopressin type 1A receptor (V1AR) expression becomes increased. Additionally, mice with cardiac-restricted V1AR overexpression develop cardiomyopathy and decreased β-adrenergic receptor (βAR) responsiveness. This led us to hypothesize that V1AR signaling regulates βAR responsiveness and in doing so contributes to development of heart failure. METHODS AND RESULTS Transaortic constriction resulted in decreased cardiac function and βAR density and increased cardiac V1AR expression, effects reversed by a V1AR-selective antagonist. Molecularly, V1AR stimulation led to decreased βAR ligand affinity, as well as βAR-induced Ca(2+) mobilization and cAMP generation in isolated adult cardiomyocytes, effects recapitulated via ex vivo Langendorff analysis. V1AR-mediated regulation of βAR responsiveness was demonstrated to occur in a previously unrecognized Gq protein-independent/G protein receptor kinase-dependent manner. CONCLUSIONS This newly discovered relationship between cardiac V1AR and βAR may be informative for the treatment of patients with acute decompensated heart failure and elevated arginine vasopressin.
Collapse
Affiliation(s)
- Douglas G Tilley
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.).
| | - Weizhong Zhu
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Valerie D Myers
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Larry A Barr
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Erhe Gao
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Xue Li
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Jianliang Song
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Rhonda L Carter
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Catherine A Makarewich
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Daohai Yu
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Constantine D Troupes
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Laurel A Grisanti
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Ryan C Coleman
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Walter J Koch
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Steven R Houser
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Joseph Y Cheung
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Arthur M Feldman
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| |
Collapse
|
56
|
See Hoe LE, Schilling JM, Tarbit E, Kiessling CJ, Busija AR, Niesman IR, Du Toit E, Ashton KJ, Roth DM, Headrick JP, Patel HH, Peart JN. Sarcolemmal cholesterol and caveolin-3 dependence of cardiac function, ischemic tolerance, and opioidergic cardioprotection. Am J Physiol Heart Circ Physiol 2014; 307:H895-903. [PMID: 25063791 DOI: 10.1152/ajpheart.00081.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cholesterol-rich caveolar microdomains and associated caveolins influence sarcolemmal ion channel and receptor function and protective stress signaling. However, the importance of membrane cholesterol content to cardiovascular function and myocardial responses to ischemia-reperfusion (I/R) and cardioprotective stimuli are unclear. We assessed the effects of graded cholesterol depletion with methyl-β-cyclodextrin (MβCD) and lifelong knockout (KO) or overexpression (OE) of caveolin-3 (Cav-3) on cardiac function, I/R tolerance, and opioid receptor (OR)-mediated protection. Langendorff-perfused hearts from young male C57Bl/6 mice were untreated or treated with 0.02-1.0 mM MβCD for 25 min to deplete membrane cholesterol and disrupt caveolae. Hearts were subjected to 25-min ischemia/45-min reperfusion, and the cardioprotective effects of morphine applied either acutely or chronically [sustained ligand-activated preconditioning (SLP)] were assessed. MβCD concentration dependently reduced normoxic contractile function and postischemic outcomes in association with graded (10-30%) reductions in sarcolemmal cholesterol. Cardioprotection with acute morphine was abolished with ≥20 μM MβCD, whereas SLP was more robust and only inhibited with ≥200 μM MβCD. Deletion of Cav-3 also reduced, whereas Cav-3 OE improved, myocardial I/R tolerance. Protection via SLP remained equally effective in Cav-3 KO mice and was additive with innate protection arising with Cav-3 OE. These data reveal the membrane cholesterol dependence of normoxic myocardial and coronary function, I/R tolerance, and OR-mediated cardioprotection in murine hearts (all declining with cholesterol depletion). In contrast, baseline function appears insensitive to Cav-3, whereas cardiac I/R tolerance parallels Cav-3 expression. Novel SLP appears unique, being less sensitive to cholesterol depletion than acute OR protection and arising independently of Cav-3 expression.
Collapse
Affiliation(s)
- Louise E See Hoe
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Emiri Tarbit
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| | - Can J Kiessling
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia; and
| | - Anna R Busija
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Ingrid R Niesman
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Eugene Du Toit
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| | - Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia; and
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California; Department of Anesthesiology, University of California, San Diego, California
| | - John P Headrick
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California; Department of Anesthesiology, University of California, San Diego, California
| | - Jason N Peart
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia;
| |
Collapse
|
57
|
Agarwal SR, Yang PC, Rice M, Singer CA, Nikolaev VO, Lohse MJ, Clancy CE, Harvey RD. Role of membrane microdomains in compartmentation of cAMP signaling. PLoS One 2014; 9:e95835. [PMID: 24752595 PMCID: PMC3994114 DOI: 10.1371/journal.pone.0095835] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 03/31/2014] [Indexed: 12/03/2022] Open
Abstract
Spatially restricting cAMP production to discrete subcellular locations permits selective regulation of specific functional responses. But exactly where and how cAMP signaling is confined is not fully understood. Different receptors and adenylyl cyclase isoforms responsible for cAMP production are not uniformly distributed between lipid raft and non-lipid raft domains of the plasma membrane. We sought to determine the role that these membrane domains play in organizing cAMP responses in HEK293 cells. The freely diffusible FRET-based biosensor Epac2-camps was used to measure global cAMP responses, while versions of the probe targeted to lipid raft (Epac2-MyrPalm) and non-raft (Epac2-CAAX) domains were used to monitor local cAMP production near the plasma membrane. Disruption of lipid rafts by cholesterol depletion selectively altered cAMP responses produced by raft-associated receptors. The results indicate that receptors associated with lipid raft as well as non-lipid raft domains can contribute to global cAMP responses. In addition, basal cAMP activity was found to be significantly higher in non-raft domains. This was supported by the fact that pharmacologic inhibition of adenylyl cyclase activity reduced basal cAMP activity detected by Epac2-CAAX but not Epac2-MyrPalm or Epac2-camps. Responses detected by Epac2-CAAX were also more sensitive to direct stimulation of adenylyl cyclase activity, but less sensitive to inhibition of phosphodiesterase activity. Quantitative modeling was used to demonstrate that differences in adenylyl cyclase and phosphodiesterase activities are necessary but not sufficient to explain compartmentation of cAMP associated with different microdomains of the plasma membrane.
Collapse
Affiliation(s)
- Shailesh R. Agarwal
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Pei-Chi Yang
- Department of Pharmacology, University of California Davis, Davis, California, United States of America
| | - Monica Rice
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Cherie A. Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Viacheslav O. Nikolaev
- European Heart Research Institute Gottingen, University of Göttingen, Göttingen, Germany
| | - Martin J. Lohse
- Department of Pharmacology, University of Würzburg, Würzburg, Germany
| | - Colleen E. Clancy
- Department of Pharmacology, University of California Davis, Davis, California, United States of America
| | - Robert D. Harvey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
58
|
Bruzzone A, Saulière A, Finana F, Sénard JM, Lüthy I, Galés C. Dosage-dependent regulation of cell proliferation and adhesion through dual β2-adrenergic receptor/cAMP signals. FASEB J 2013; 28:1342-54. [PMID: 24308976 DOI: 10.1096/fj.13-239285] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of β-adrenergic receptors (β-ARs) remains controversial in normal and tumor breast. Herein we explore the cAMP signaling involved in β-AR-dependent control of proliferation and adhesion of nontumor human breast cell line MCF-10A. Low concentrations of a β-agonist, isoproterenol (ISO), promote cell adhesion (87.5% cells remaining adherent to the plastic dishes following specific detachment vs. 35.0% in control, P<0.001), while increasing concentrations further engages an additional 36% inhibition of Erk1/2 phosphorylation (p-Erk1/2)-dependent cell proliferation (P<0.01). Isoproterenol dose response on cell adhesion was fitted to a 2-site curve (EC50(1): 16.5±11.5 fM, EC50(2): 4.08±3.09 nM), while ISO significantly inhibited p-Erk1/2 according to a 1-site model (EC50: 0.25±0.13 nM). Using β-AR-selective agonist/antagonists and cAMP analogs/inhibitors, we identified a dosage-dependent signaling in which low ISO concentrations target a β2-AR population localized in raft microdomains and stimulate a Gs/cAMP/Epac/adhesion-signaling module, while higher concentrations engage a concomitant activation of another β2-AR population outside rafts and inhibit the proliferation by a Gs/cAMP/PKA-dependent signaling module. Our data provide a new molecular basis for the dose-dependent switch of β-AR signaling. This study also sheds light on a new cAMP pathway core mechanism with a single receptor triggering dual cAMP signaling controlled by PKA or Epac but with different cellular outputs.
Collapse
Affiliation(s)
- Ariana Bruzzone
- 2Inserm U1048, Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, 1, avenue Jean-Poulhès, BP84225, 31432 Toulouse Cedex 4, France.
| | | | | | | | | | | |
Collapse
|
59
|
Kumari R, Castillo C, Francesconi A. Agonist-dependent signaling by group I metabotropic glutamate receptors is regulated by association with lipid domains. J Biol Chem 2013; 288:32004-19. [PMID: 24045944 DOI: 10.1074/jbc.m113.475863] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Group I metabotropic glutamate receptors (mGluRs), mGluR1 and mGluR5, play critical functions in forms of activity-dependent synaptic plasticity and synapse remodeling in physiological and pathological states. Importantly, in animal models of fragile X syndrome, group I mGluR activity is abnormally enhanced, a dysfunction that may partly underlie cognitive deficits in the condition. Lipid rafts are cholesterol- and sphingolipid-enriched membrane domains that are thought to form transient signaling platforms for ligand-activated receptors. Many G protein-coupled receptors, including group I mGluRs, are present in lipid rafts, but the mechanisms underlying recruitment to these membrane domains remain incompletely understood. Here, we show that mGluR1 recruitment to lipid rafts is enhanced by agonist binding and is supported at least in part by an intact cholesterol recognition/interaction amino acid consensus (CRAC) motif in the receptor. Substitutions of critical residues in the motif reduce mGluR1 association with lipid rafts and agonist-induced, mGluR1-dependent activation of extracellular-signal-activated kinase1/2 MAP kinase (ERK-MAPK). We find that alteration of membrane cholesterol content or perturbation of lipid rafts regulates agonist-dependent activation of ERK-MAPK by group I mGluRs, suggesting a potential function for cholesterol as a positive allosteric modulator of receptor function(s). Together, these findings suggest that drugs that alter membrane cholesterol levels or directed to the receptor-cholesterol interface could be employed to modulate abnormal group I mGluR activity in neuropsychiatric conditions, including fragile X syndrome.
Collapse
Affiliation(s)
- Ranju Kumari
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | | |
Collapse
|
60
|
Khelashvili G, LeVine MV, Shi L, Quick M, Javitch JA, Weinstein H. The membrane protein LeuT in micellar systems: aggregation dynamics and detergent binding to the S2 site. J Am Chem Soc 2013; 135:14266-75. [PMID: 23980525 PMCID: PMC3788620 DOI: 10.1021/ja405984v] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Structural and functional properties of integral membrane proteins are often studied in detergent micellar environments (proteomicelles), but how such proteomicelles form and organize is not well understood. This makes it difficult to evaluate the relationship between the properties of the proteins measured in such a detergent-solubilized form and under native conditions. To obtain mechanistic information about this relationship for the leucine transporter (LeuT), a prokaryotic homologue of the mammalian neurotransmitter/sodium symporters (NSSs), we studied the properties of proteomicelles formed by n-dodecyl-β,D-maltopyranoside (DDM) detergent. Extensive atomistic molecular dynamics simulations of different protein/detergent/water number ratios revealed the formation of a proteomicelle characterized by a constant-sized shell of detergents surrounding LeuT protecting its transmembrane segments from unfavorable hydrophobic/hydrophilic exposure. Regardless of the DDM content in the simulated system, this shell consisted of a constant number of DDM molecules (∼120 measured at a 4 Å cutoff distance from LeuT). In contrast, the overall number of DDMs in the proteomicelle (aggregation number) was found to depend on the detergent concentration, reaching a saturation value of 226±17 DDMs in the highest concentration regime simulated. Remarkably, we found that at high detergent-to-protein ratios we observed two independent ways of DDM penetration into LeuT, both leading to a positioning of the DDM molecule in the second substrate (S2) binding site of LeuT. Consonant with several recent experimental studies demonstrating changes in functional properties of membrane proteins due to detergent, our findings highlight how the environment in which the membrane proteins are examined may affect the outcome and interpretation of their mechanistic features.
Collapse
Affiliation(s)
- George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University (WCMC) , New York, New York 10065, United States
| | | | | | | | | | | |
Collapse
|
61
|
Ultra-LAMA, ultra-LABA, ultra-inhaled steroids? The future has landed. Arch Bronconeumol 2013; 49:131-4. [PMID: 23415574 DOI: 10.1016/j.arbres.2012.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 11/12/2012] [Accepted: 11/28/2012] [Indexed: 11/24/2022]
|
62
|
Cang X, Du Y, Mao Y, Wang Y, Yang H, Jiang H. Mapping the Functional Binding Sites of Cholesterol in β2-Adrenergic Receptor by Long-Time Molecular Dynamics Simulations. J Phys Chem B 2013; 117:1085-94. [DOI: 10.1021/jp3118192] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaohui Cang
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yun Du
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yanyan Mao
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yuanyuan Wang
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Huaiyu Yang
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Hualiang Jiang
- Drug Discovery and
Design Center, State Key Laboratory of Drug Research, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
63
|
Ibrahim S, McCartney A, Markosyan N, Smyth EM. Heterodimerization with the prostacyclin receptor triggers thromboxane receptor relocation to lipid rafts. Arterioscler Thromb Vasc Biol 2012; 33:60-6. [PMID: 23162015 DOI: 10.1161/atvbaha.112.300536] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Prostacyclin and thromboxane mediate opposing cardiovascular actions through receptors termed IP and TP, respectively. When dimerized with IP, the TP shifts to IP-like function. IP localizes to cholesterol-enriched membrane rafts, but TP and IPTP heterodimer localization is not defined. We examined these receptors' membrane localization and the role of rafts in receptor function. METHODS AND RESULTS Microdomain distribution of IP, TP, and IPTP heterodimers was examined in COS-7 cells by measuring energy transfer from renilla luciferase-fused receptors to fluorescently labeled rafts. IP raft association was confirmed. TP was raft excluded, but redistributed to rafts upon dimerization with IP. Signaling of the IP and IPTP heterodimer, but not TP alone, was suppressed after raft disruption by cholesterol depletion. Cholesterol enrichment also selectively suppressed IP and IPTP function. Native IP and IPTP signaling in smooth muscle cells and macrophages were similarly sensitive to cholesterol manipulation, whereas macrophages from hypercholesterolemic mice displayed suppressed IP and IPTP function. CONCLUSIONS IP and TP function within distinct microdomains. Raft incorporation of TP in the IPTP heterodimer likely facilitates its signaling shift. We speculate that changes in IP and IPTP signaling after perturbation of membrane cholesterol may contribute to cardiovascular disease associated with hypercholesterolemia.
Collapse
Affiliation(s)
- Salam Ibrahim
- Institute for Translational Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
64
|
Ma L, Dong F, Zaid M, Kumar A, Zha X. ABCA1 protein enhances Toll-like receptor 4 (TLR4)-stimulated interleukin-10 (IL-10) secretion through protein kinase A (PKA) activation. J Biol Chem 2012; 287:40502-12. [PMID: 23055522 DOI: 10.1074/jbc.m112.413245] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND ABCA1 is known to suppress proinflammatory cytokines. RESULTS ABCA1 activates PKA and up-regulates anti-inflammatory cytokine IL-10. Elevated PKA transforms macrophages to M2-like phenotype. Disrupting lipid rafts by statins MCD, and filipin recuperates ABCA1 phenotype and likely functions downstream of ABCA1. CONCLUSION By modulating cholesterol, ABCA1 activates PKA. This generates M2-like macrophages. SIGNIFICANCE ABCA1 does not simply suppress inflammatory response. It promotes M2-like activation and facilitates resolution. Nonresolving inflammatory response from macrophages is a major characteristic of atherosclerosis. Macrophage ABCA1 has been previously shown to suppress the secretion of proinflammatory cytokine. In the present study, we demonstrate that ABCA1 also promotes the secretion of IL-10, an anti-inflammatory cytokine critical for inflammation resolution. ABCA1(+/+) bone marrow-derived macrophages secrete more IL-10 but less proinflammatory cytokines than ABCA1(-/-) bone marrow-derived macrophages, similar to alternatively activated (M2) macrophages. We present evidence that ABCA1 activates PKA and that this elevated PKA activity contributes to M2-like inflammatory response from ABCA1(+/+) bone marrow-derived macrophages. Furthermore, cholesterol lowering by statins, methyl-β-cyclodextrin, or filipin also activates PKA and, consequently, transforms macrophages toward M2-like phenotype. Conversely, cholesterol enrichment suppresses PKA activity and promotes M1-like inflammatory response. As the primary function of ABCA1 is cholesterol removal, our results suggest that ABCA1 activates PKA by regulating cholesterol. Indeed, forced cholesterol enrichment in ABCA1-expressing macrophages suppresses PKA activation and elicits M1-like response. Collectively, these findings reveal a novel protective process by ABCA1-activated PKA in macrophages. They also suggest cholesterol lowering in extra-hepatic tissues by statins as an anti-inflammation strategy.
Collapse
Affiliation(s)
- Loretta Ma
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
65
|
Sato M, Hutchinson DS, Halls ML, Furness SGB, Bengtsson T, Evans BA, Summers RJ. Interaction with caveolin-1 modulates G protein coupling of mouse β3-adrenoceptor. J Biol Chem 2012; 287:20674-88. [PMID: 22535965 DOI: 10.1074/jbc.m111.280651] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caveolins act as scaffold proteins in multiprotein complexes and have been implicated in signaling by G protein-coupled receptors. Studies using knock-out mice suggest that β(3)-adrenoceptor (β(3)-AR) signaling is dependent on caveolin-1; however, it is not known whether caveolin-1 is associated with the β(3)-AR or solely with downstream signaling proteins. We have addressed this question by examining the impact of membrane rafts and caveolin-1 on the differential signaling of mouse β(3a)- and β(3b)-AR isoforms that diverge at the distal C terminus. Only the β(3b)-AR promotes pertussis toxin (PTX)-sensitive cAMP accumulation. When cells expressing the β(3a)-AR were treated with filipin III to disrupt membrane rafts or transfected with caveolin-1 siRNA, the cyclic AMP response to the β(3)-AR agonist CL316243 became PTX-sensitive, suggesting Gα(i/o) coupling. The β(3a)-AR C terminus, SP(384)PLNRF(389)DGY(392)EGARPF(398)PT, resembles a caveolin interaction motif. Mutant β(3a)-ARs (F389A/Y392A/F398A or P384S/F389A) promoted PTX-sensitive cAMP responses, and in situ proximity assays demonstrated an association between caveolin-1 and the wild type β(3a)-AR but not the mutant receptors. In membrane preparations, the β(3b)-AR activated Gα(o) and mediated PTX-sensitive cAMP responses, whereas the β(3a)-AR did not activate Gα(i/o) proteins. The endogenous β(3a)-AR displayed Gα(i/o) coupling in brown adipocytes from caveolin-1 knock-out mice or in wild type adipocytes treated with filipin III. Our studies indicate that interaction of the β(3a)-AR with caveolin inhibits coupling to Gα(i/o) proteins and suggest that signaling is modulated by a raft-enriched complex containing the β(3a)-AR, caveolin-1, Gα(s), and adenylyl cyclase.
Collapse
Affiliation(s)
- Masaaki Sato
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and the Department of Pharmacology, Monash University, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
66
|
Lohse MJ, Nuber S, Hoffmann C. Fluorescence/bioluminescence resonance energy transfer techniques to study G-protein-coupled receptor activation and signaling. Pharmacol Rev 2012; 64:299-336. [PMID: 22407612 DOI: 10.1124/pr.110.004309] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fluorescence and bioluminescence resonance energy transfer (FRET and BRET) techniques allow the sensitive monitoring of distances between two labels at the nanometer scale. Depending on the placement of the labels, this permits the analysis of conformational changes within a single protein (for example of a receptor) or the monitoring of protein-protein interactions (for example, between receptors and G-protein subunits). Over the past decade, numerous such techniques have been developed to monitor the activation and signaling of G-protein-coupled receptors (GPCRs) in both the purified, reconstituted state and in intact cells. These techniques span the entire spectrum from ligand binding to the receptors down to intracellular second messengers. They allow the determination and the visualization of signaling processes with high temporal and spatial resolution. With these techniques, it has been demonstrated that GPCR signals may show spatial and temporal patterning. In particular, evidence has been provided for spatial compartmentalization of GPCRs and their signals in intact cells and for distinct physiological consequences of such spatial patterning. We review here the FRET and BRET technologies that have been developed for G-protein-coupled receptors and their signaling proteins (G-proteins, effectors) and the concepts that result from such experiments.
Collapse
Affiliation(s)
- Martin J Lohse
- Institute of Pharmacology and Toxicology, Versbacher Str. 9, 97078 Würzburg, Germany.
| | | | | |
Collapse
|
67
|
Oddi S, Dainese E, Sandiford S, Fezza F, Lanuti M, Chiurchiù V, Totaro A, Catanzaro G, Barcaroli D, De Laurenzi V, Centonze D, Mukhopadhyay S, Selent J, Howlett AC, Maccarrone M. Effects of palmitoylation of Cys(415) in helix 8 of the CB(1) cannabinoid receptor on membrane localization and signalling. Br J Pharmacol 2012; 165:2635-51. [PMID: 21895628 PMCID: PMC3423250 DOI: 10.1111/j.1476-5381.2011.01658.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 07/15/2011] [Accepted: 08/05/2011] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The CB(1) cannabinoid receptor is regulated by its association with membrane microdomains such as lipid rafts. Here, we investigated the role of palmitoylation of the CB(1) receptor by analysing the functional consequences of site-specific mutation of Cys(415) , the likely site of palmitoylation at the end of helix 8, in terms of membrane association, raft targeting and signalling. EXPERIMENTAL APPROACH The palmitoylation state of CB(1) receptors in rat forebrain was assessed by depalmitoylation/repalmitoylation experiments. Cys(415) was replaced with alanine by site-directed mutagenesis. Green fluorescence protein chimeras of both wild-type and mutant receptors were transiently expressed and functionally characterized in SH-SY5Y cells and HEK-293 cells by means of confocal microscopy, cytofluorimetry and competitive binding assays. Confocal fluorescence recovery after photobleaching was used to assess receptor membrane dynamics, whereas signalling activity was assessed by [(35) S]GTPγS, cAMP and co-immunoprecipitation assays. KEY RESULTS Endogenous CB(1) receptors in rat brain were palmitoylated. Mutation of Cys(415) prevented the palmitoylation of the receptor in transfected cells and reduced its recruitment to plasma membrane and lipid rafts; it also increased protein diffusional mobility. The same mutation markedly reduced the functional coupling of CB(1) receptors with G-proteins and adenylyl cyclase, whereas depalmitoylation abolished receptor association with a specific subset of G-proteins. CONCLUSIONS AND IMPLICATIONS CB(1) receptors were post-translationally modified by palmitoylation. Mutation of Cys(415) provides a receptor that is functionally impaired in terms of membrane targeting and signalling. LINKED ARTICLES This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.
Collapse
Affiliation(s)
- Sergio Oddi
- Department of Biomedical Sciences, University of TeramoTeramo, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Enrico Dainese
- Department of Biomedical Sciences, University of TeramoTeramo, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Simone Sandiford
- Neuroscience/Drug Abuse Research Program, Biomedical Biotechnology Research Institute, North Carolina Central UniversityDurham, NC, USA
| | - Filomena Fezza
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
- Department of Experimental Medicine and Biochemical Sciences, University of Rome ‘Tor Vergata’Rome, Italy
| | - Mirko Lanuti
- Department of Biomedical Sciences, University of TeramoTeramo, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Valerio Chiurchiù
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Antonio Totaro
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Giuseppina Catanzaro
- Department of Biomedical Sciences, University of TeramoTeramo, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| | - Daniela Barcaroli
- Department of Biomedical Sciences, University of Chieti-Pescara ‘G. d'Annunzio’Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Biomedical Sciences, University of Chieti-Pescara ‘G. d'Annunzio’Chieti, Italy
| | - Diego Centonze
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
- Department of Neurosciences, University of Rome ‘Tor Vergata’Rome, Italy
| | - Somnath Mukhopadhyay
- Neuroscience/Drug Abuse Research Program, Biomedical Biotechnology Research Institute, North Carolina Central UniversityDurham, NC, USA
| | - Jana Selent
- Research Group of biomedical Informatics (GRIB-IMIM), University of Pompeu Fabra, Barcelona Biomedical Research Park (PRBB)Barcelona, Spain
| | - Allyn C Howlett
- Department of Physiology and Pharmacology, Wake Forest University Health SciencesWinston-Salem, NC, USA
| | - Mauro Maccarrone
- Department of Biomedical Sciences, University of TeramoTeramo, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation I.R.C.C.S.Rome, Italy
| |
Collapse
|
68
|
Scarselli M, Annibale P, Radenovic A. Cell type-specific β2-adrenergic receptor clusters identified using photoactivated localization microscopy are not lipid raft related, but depend on actin cytoskeleton integrity. J Biol Chem 2012; 287:16768-80. [PMID: 22442147 DOI: 10.1074/jbc.m111.329912] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent developments in the field of optical super-resolution techniques allow both a 10-fold increase in resolution as well as an increased ability to quantify the number of labeled molecules visualized in the fluorescence measurement. By using photoactivated localization microscopy (PALM) and an experimental approach based on the systematic comparison with a nonclustering peptide as a negative control, we found that the prototypical G protein-coupled receptor β2-adrenergic receptor is partially preassociated in nanoscale-sized clusters only in the cardiomyocytes, such as H9C2 cells, but not in other cell lines, such as HeLa and Chinese hamster ovary (CHO). The addition of the agonist for very short times or the addition of the inverse agonist did not significantly affect the organization of receptor assembly. To investigate the mechanism governing cluster formation, we altered plasma membrane properties with cholesterol removal and actin microfilament disruption. Although cholesterol is an essential component of cell membranes and it is supposed to be enriched in the lipid rafts, its sequestration and removal did not affect receptor clustering, whereas the inhibition of actin polymerization did decrease the number of clusters. Our findings are therefore consistent with a model in which β2 receptor clustering is influenced by the actin cytoskeleton, but it does not rely on lipid raft integrity, thus ruling out the possibility that cell type-specific β2 receptor clustering is associated with the raft.
Collapse
Affiliation(s)
- Marco Scarselli
- Laboratory of Nanoscale Biology, Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
69
|
Jiang X, Pan H, Nabhan JF, Krishnan R, Koziol-White C, Panettieri RA, Lu Q. A novel EST-derived RNAi screen reveals a critical role for farnesyl diphosphate synthase in β2-adrenergic receptor internalization and down-regulation. FASEB J 2012; 26:1995-2007. [PMID: 22278941 DOI: 10.1096/fj.11-193870] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The β2-adrenergic receptor (β2AR) plays important physiological roles in the heart and lung and is the primary target of β-agonists, the mainstay asthma drugs. Activation of β2AR by β-agonists is attenuated by receptor down-regulation, which ensures transient stimulation of the receptor but reduces the efficacy of β-agonists. Here we report the identification, through a functional genome-wide RNA interference (RNAi) screen, of new genes critically involved in β2AR down-regulation. We developed a lentivirus-based RNAi library consisting of 26-nt short-hairpin RNAs (shRNAs). The library was generated enzymatically from a large collection of expressed sequence tag (EST) DNAs corresponding to ∼20,000 human genes and contains on average ∼6 highly potent shRNAs (>75% knockdown efficiency) for each gene. Using this novel shRNA library, together with a robust cell model for β2AR expression, we performed fluorescence-activated cell sorting and isolated cells that, as a consequence of shRNA-mediated gene inactivation, exhibited defective agonist-induced down-regulation. The screen discovered several previously unrecognized β2AR regulators, including farnesyl diphosphate synthase (FDPS). We showed that inactivation of FDPS by shRNA, small interfering RNA, or the highly specific pharmaceutical inhibitor alendronate inhibited β2AR down-regulation. Notably, in human airway smooth muscle cells, the physiological target of β-agonists, alendronate treatment functionally reversed agonist-induced endogenous β2AR loss as indicated by an increase in cAMP production. FDPS inactivation interfered with β2AR internalization into endosomes through disrupting the membrane localization of the Rab5 small GTPase. Furthermore, Rab5 overexpression reversed the deficient receptor down-regulation induced by alendronate, suggesting that FDPS regulates receptor down-regulation in a Rab5-dependent manner. Together, our findings reveal a FDPS-dependent mechanism in the internalization and down-regulation of β2AR, identify FDPS as a potential target for improving the therapeutic efficacy of β-agonists, and demonstrate the utility of the unique EST-derived shRNA library for functional genetics studies.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- Program in Molecular and Integrative Physiological Sciences, Harvard School of Public Health, 665 Huntington Ave., Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
70
|
Cambi A, Lidke DS. Nanoscale membrane organization: where biochemistry meets advanced microscopy. ACS Chem Biol 2012; 7:139-49. [PMID: 22004174 DOI: 10.1021/cb200326g] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding the molecular mechanisms that shape an effective cellular response is a fundamental question in biology. Biochemical measurements have revealed critical information about the order of protein-protein interactions along signaling cascades but lack the resolution to determine kinetics and localization of interactions on the plasma membrane. Furthermore, the local membrane environment influences membrane receptor distributions and dynamics, which in turn influences signal transduction. To measure dynamic protein interactions and elucidate the consequences of membrane architecture interplay, direct measurements at high spatiotemporal resolution are needed. In this review, we discuss the biochemical principles regulating membrane nanodomain formation and protein function, ranging from the lipid nanoenvironment to the cortical cytoskeleton. We also discuss recent advances in fluorescence microscopy that are making it possible to quantify protein organization and biochemical events at the nanoscale in the living cell membrane.
Collapse
Affiliation(s)
- Alessandra Cambi
- Department of Tumor Immunology,
Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Diane S. Lidke
- Department of Pathology and
Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States
| |
Collapse
|
71
|
Yorgancioglu A. Indacaterol in chronic obstructive pulmonary disease: an update for clinicians. Ther Adv Chronic Dis 2012; 3:25-36. [PMID: 23251766 PMCID: PMC3513896 DOI: 10.1177/2040622311426204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchodilation is the cornerstone of chronic obstructive pulmonary disease (COPD) management and is based on regular treatment with one or more long-acting β(2) agonists (LABAs). A novel bronchodilator, indacaterol, satisfies the requirements of an efficacious LABA: it has a relatively longer duration of action compared with existing LABAs and a fast onset of action. This review is a presentation of data on indacaterol with respect to its molecular characteristics as well as comparisons with other long-acting bronchodilators. Data from 12 relevant trials show that once-daily indacaterol provides significant, consistent and clinically important improvements in lung function (forced expiratory volume in 1 second), significant improvements in breathlessness and health status at least as good as or better than tiotropium, salmeterol and formoterol, and reduction in requirement for relief medication compared with tiotropium, salmeterol and formoterol.
Collapse
|
72
|
Desai AJ, Miller LJ. Sensitivity of cholecystokinin receptors to membrane cholesterol content. Front Endocrinol (Lausanne) 2012; 3:123. [PMID: 23087674 PMCID: PMC3475150 DOI: 10.3389/fendo.2012.00123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Accepted: 10/01/2012] [Indexed: 12/18/2022] Open
Abstract
Cholesterol represents a structurally and functionally important component of the eukaryotic cell membrane, where it increases lipid order, affects permeability, and influences the lateral mobility and conformation of membrane proteins. Several G protein-coupled receptors have been shown to be affected by the cholesterol content of the membrane, with functional impact on their ligand binding and signal transduction characteristics. The effects of cholesterol can be mediated directly by specific molecular interactions with the receptor and/or indirectly by altering the physical properties of the membrane. This review focuses on the importance and differential effects of membrane cholesterol on the activity of cholecystokinin (CCK) receptors. The type 1 CCK receptor is quite sensitive to its cholesterol environment, while the type 2 CCK receptor is not. The possible structural basis for this differential impact is explored and the implications of pathological states, such as metabolic syndrome, in which membrane cholesterol may be increased and CCK1R function may be abnormal are discussed. This is believed to have substantial potential importance for the development of drugs targeting the CCK receptor.
Collapse
Affiliation(s)
| | - Laurence J. Miller
- *Correspondence: Laurence J. Miller, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259, USA. e-mail:
| |
Collapse
|
73
|
Insel PA. β(2)-Adrenergic receptor polymorphisms and signaling: Do variants influence the "memory" of receptor activation? Sci Signal 2011; 4:pe37. [PMID: 21868355 DOI: 10.1126/scisignal.2002352] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nonsynonymous, coding sequence single-nucleotide polymorphisms in β(2)-adrenergic receptors were first recognized almost 20 years ago, but a full understanding of their impact on signal transduction-especially on receptor abundance in native cells and their clinical importance-remains unclear. New evidence has revealed a feature of the Arg(16)Gly variant of β(2)-adrenergic receptors that has not been previously noted: a difference in the rate of response upon repeated stimulation of the receptors, such that the Arg(16) variant shows slower activation and the Gly(16) variant faster activation of cyclic adenosine monophosphate (cAMP) formation-a feature that the authors term "receptor memory." This is an intriguing idea but will require confirmation and demonstration of its functional importance in vivo and its possible contribution to clinical responses, especially in terms of the administration of β(2)-adrenergic agonists.
Collapse
Affiliation(s)
- Paul A Insel
- Departments of Pharmacology and Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
74
|
Pontier SM, Schweisguth F. Glycosphingolipids in signaling and development: From liposomes to model organisms. Dev Dyn 2011; 241:92-106. [DOI: 10.1002/dvdy.22766] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2011] [Indexed: 01/05/2023] Open
|
75
|
Potter RM, Harikumar KG, Wu SV, Miller LJ. Differential sensitivity of types 1 and 2 cholecystokinin receptors to membrane cholesterol. J Lipid Res 2011; 53:137-48. [PMID: 22021636 DOI: 10.1194/jlr.m020065] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies indicate that membrane cholesterol can associate with G protein-coupled receptors (GPCRs) and affect their function. Previously, we reported that manipulation of membrane cholesterol affects ligand binding and signal transduction of the type 1 cholecystokinin receptor (CCK1R), a Class A GPCR. We now demonstrate that the closely related type 2 cholecystokinin receptor (CCK2R) does not share this cholesterol sensitivity. The sequences of both receptors reveal almost identical cholesterol interaction motifs in analogous locations in transmembrane segments two, three, four, and five. The disparity in cholesterol sensitivity between these receptors, despite their close structural relationship, provides a unique opportunity to define the possible structural basis of cholesterol sensitivity of CCK1R. To evaluate the relative contributions of different regions of CCK1R to cholesterol sensitivity, we performed ligand binding studies and biological activity assays of wild-type and CCK2R/CCK1R chimeric receptor-bearing Chinese hamster ovary cells after manipulation of membrane cholesterol. We also extended these studies to site-directed mutations within the cholesterol interaction motifs. The results contribute to a better understanding of the structural requirements for cholesterol sensitivity in CCK1R and provides insight into the function of other cholesterol-sensitive Class A GPCRs.
Collapse
Affiliation(s)
- Ross M Potter
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | | | | |
Collapse
|
76
|
PI3K/Akt signaling requires spatial compartmentalization in plasma membrane microdomains. Proc Natl Acad Sci U S A 2011; 108:14509-14. [PMID: 21873248 DOI: 10.1073/pnas.1019386108] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Spatial compartmentalization of signaling pathway components generally defines the specificity and enhances the efficiency of signal transduction. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is known to be compartmentalized within plasma membrane microdomains; however, the underlying mechanisms and functional impact of this compartmentalization are not well understood. Here, we show that phosphoinositide-dependent kinase 1 is activated in membrane rafts in response to growth factors, whereas the negative regulator of the pathway, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), is primarily localized in nonraft regions. Alteration of this compartmentalization, either by genetic targeting or ceramide-induced recruitment of PTEN to rafts, abolishes the activity of the entire pathway. These findings reveal critical steps in raft-mediated PI3K/Akt activation and demonstrate the essential role of membrane microdomain compartmentalization in enabling PI3K/Akt signaling. They further suggest that dysregulation of this compartmentalization may underlie pathological complications such as insulin resistance.
Collapse
|
77
|
Maccarrone M, Bernardi G, Agrò AF, Centonze D. Cannabinoid receptor signalling in neurodegenerative diseases: a potential role for membrane fluidity disturbance. Br J Pharmacol 2011; 163:1379-90. [PMID: 21323908 PMCID: PMC3165948 DOI: 10.1111/j.1476-5381.2011.01277.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/13/2011] [Accepted: 01/24/2011] [Indexed: 11/30/2022] Open
Abstract
Type-1 cannabinoid receptor (CB(1)) is the most abundant G-protein-coupled receptor (GPCR) in the brain. CB(1) and its endogenous agonists, the so-called 'endocannabinoids (eCBs)', belong to an ancient neurosignalling system that plays important functions in neurodegenerative and neuroinflammatory disorders like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. For this reason, research on the therapeutic potential of drugs modulating the endogenous tone of eCBs is very intense. Several GPCRs reside within subdomains of the plasma membranes that contain high concentrations of cholesterol: the lipid rafts. Here, the hypothesis that changes in membrane fluidity alter function of the endocannabinoid system, as well as progression of particular neurodegenerative diseases, is described. To this end, the impact of membrane cholesterol on membrane properties and hence on neurodegenerative diseases, as well as on CB(1) signalling in vitro and on CB(1) -dependent neurotransmission within the striatum, is discussed. Overall, present evidence points to the membrane environment as a critical regulator of signal transduction triggered by CB(1) , and calls for further studies aimed at better clarifying the contribution of membrane lipids to eCBs signalling. The results of these investigations might be exploited also for the development of novel therapeutics able to combat disorders associated with abnormal activity of CB(1).
Collapse
Affiliation(s)
- M Maccarrone
- Department of Biomedical Sciences, University of Teramo, Teramo 64100, Italy.
| | | | | | | |
Collapse
|
78
|
Norambuena A, Schwartz MA. Effects of integrin-mediated cell adhesion on plasma membrane lipid raft components and signaling. Mol Biol Cell 2011; 22:3456-64. [PMID: 21795400 PMCID: PMC3172269 DOI: 10.1091/mbc.e11-04-0361] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Anchorage dependence of cell growth, which is mediated by multiple integrin-regulated signaling pathways, is a key defense against cancer metastasis. Detachment of cells from the extracellular matrix triggers caveolin-1-dependent internalization of lipid raft components, which mediates suppression of Rho GTPases, Erk, and phosphatidylinositol 3-kinase in suspended cells. Elevation of cyclic adenosine monophosphate (cAMP) following cell detachment is also implicated in termination of growth signaling in suspended cells. Studies of integrins and lipid rafts, however, examined mainly ganglioside GM1 and glycosylphosphatidylinositol-linked proteins as lipid raft markers. In this study, we examine a wider range of lipid raft components. Whereas many raft components internalized with GM1 following cell detachment, flotillin2, connexin43, and Gα(s) remained in the plasma membrane. Loss of cell adhesion caused movement of many components from the lipid raft to the nonraft fractions on sucrose gradients, although flotillin2, connexin43, and H-Ras were resistant. Gα(s) lost its raft association, concomitant with cAMP production. Modification of the lipid tail of Gα(s) to increase its association with ordered domains blocked the detachment-induced increase in cAMP. These data define the effects of that integrin-mediated adhesion on the localization and behavior of a variety of lipid raft components and reveal the mechanism of the previously described elevation of cAMP after cell detachment.
Collapse
Affiliation(s)
- Andrés Norambuena
- Robert M. Berne Cardiovascular Research Center, Mellon Urological Cancer Research Institute, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
79
|
Valentine CD, Haggie PM. Confinement of β(1)- and β(2)-adrenergic receptors in the plasma membrane of cardiomyocyte-like H9c2 cells is mediated by selective interactions with PDZ domain and A-kinase anchoring proteins but not caveolae. Mol Biol Cell 2011; 22:2970-82. [PMID: 21680711 PMCID: PMC3154891 DOI: 10.1091/mbc.e11-01-0034] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The presence of stable multiprotein complexes containing adrenergic receptors is verified in live H9c2 cardiomyocyte-like cells by single-particle tracking. The immobilization of β-adrenergic receptors presumably contributes to the specificity of cardiac adrenergic responses. The sympathetic nervous system regulates cardiac output by activating adrenergic receptors (ARs) in cardiac myocytes. The predominant cardiac ARs, β1- and β2AR, are structurally similar but mediate distinct signaling responses. Scaffold protein–mediated compartmentalization of ARs into discrete, multiprotein complexes has been proposed to dictate differential signaling responses. To test the hypothesis that βARs integrate into complexes in live cells, we measured receptor diffusion and interactions by single-particle tracking. Unstimulated β1- and β2AR were highly confined in the membrane of H9c2 cardiomyocyte-like cells, indicating that receptors are tethered and presumably integrated into protein complexes. Selective disruption of interactions with postsynaptic density protein 95/disks large/zonula occludens-1 (PDZ)–domain proteins and A-kinase anchoring proteins (AKAPs) increased receptor diffusion, indicating that these scaffold proteins participate in receptor confinement. In contrast, modulation of interactions between the putative scaffold caveolae and β2AR did not alter receptor dynamics, suggesting that these membrane domains are not involved in β2AR confinement. For both β1- and β2AR, the receptor carboxy-terminus was uniquely responsible for scaffold interactions. Our data formally demonstrate that distinct and stable protein complexes containing β1- or β2AR are formed in the plasma membrane of cardiomyocyte-like cells and that selective PDZ and AKAP interactions are responsible for the integration of receptors into complexes.
Collapse
Affiliation(s)
- Cathleen D Valentine
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
80
|
Breton B, Sauvageau É, Zhou J, Bonin H, Le Gouill C, Bouvier M. Multiplexing of multicolor bioluminescence resonance energy transfer. Biophys J 2011; 99:4037-46. [PMID: 21156147 DOI: 10.1016/j.bpj.2010.10.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/30/2010] [Accepted: 10/15/2010] [Indexed: 10/18/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is increasingly being used to monitor protein-protein interactions and cellular events in cells. However, the ability to monitor multiple events simultaneously is limited by the spectral properties of the existing BRET partners. Taking advantage of newly developed Renilla luciferases and blue-shifted fluorescent proteins (FPs), we explored the possibility of creating novel BRET configurations using a single luciferase substrate and distinct FPs. Three new (to our knowledge) BRET assays leading to distinct color bioluminescence emission were generated and validated. The spectral properties of two of the FPs used (enhanced blue (EB) FP2 and mAmetrine) and the selection of appropriate detection filters permitted the concomitant detection of two independent BRET signals, without cross-interference, in the same cells after addition of a unique substrate for Renilla luciferase-II, coelentrazine-400a. Using individual BRET-based biosensors to monitor the interaction between G-protein-coupled receptors and G-protein subunits or activation of different G-proteins along with the production of a second messenger, we established the proof of principle that two new BRET configurations can be multiplexed to simultaneously monitor two dependent or independent cellular events. The development of this new multiplexed BRET configuration opens the way for concomitant monitoring of various independent biological processes in living cells.
Collapse
Affiliation(s)
- Billy Breton
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Canada
| | | | | | | | | | | |
Collapse
|
81
|
Vidi PA, Ejendal KF, Przybyla JA, Watts VJ. Fluorescent protein complementation assays: new tools to study G protein-coupled receptor oligomerization and GPCR-mediated signaling. Mol Cell Endocrinol 2011; 331:185-93. [PMID: 20654687 PMCID: PMC2990800 DOI: 10.1016/j.mce.2010.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 06/07/2010] [Accepted: 07/13/2010] [Indexed: 11/29/2022]
Abstract
G protein-coupled receptor (GPCR) signaling is mediated by protein-protein interactions at multiple levels. The characterization of the corresponding protein complexes is therefore paramount to the basic understanding of GPCR-mediated signal transduction. The number of documented interactions involving GPCRs is rapidly growing, and appreciating the functional significance of these complexes is clearly the next challenge. New experimental approaches including protein complementation assays (PCAs) have recently been used to examine the composition, plasma membrane targeting, and desensitization of protein complexes involved in GPCR signaling. These methods also hold promise for better understanding of drug-induced effects on GPCR interactions. This review focuses on the application of fluorescent PCAs for the study of GPCR signaling. Potential applications of PCAs in high-content screens are also presented. Non-fluorescent PCA techniques as well as combined assays for the detection of ternary and quaternary protein complexes are briefly discussed.
Collapse
Affiliation(s)
- Pierre-Alexandre Vidi
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University
| | - Karin F.K. Ejendal
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University
| | - Julie A. Przybyla
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University
| | - Val J. Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University
| |
Collapse
|
82
|
Breit A, Büch TRH, Boekhoff I, Solinski HJ, Damm E, Gudermann T. Alternative G protein coupling and biased agonism: new insights into melanocortin-4 receptor signalling. Mol Cell Endocrinol 2011; 331:232-40. [PMID: 20674667 DOI: 10.1016/j.mce.2010.07.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 05/06/2010] [Accepted: 07/13/2010] [Indexed: 11/20/2022]
Abstract
The melanocortin-4 receptor (MC4R) is a prototypical G protein-coupled receptor (GPCR) that plays a considerable role in controlling appetite and energy homeostasis. Signalling initiated by MC4R is orchestrated by multiple agonists, inverse agonism and by interactions with accessory proteins. The exact molecular events translating MC4R signalling into its physiological role, however, are not fully understood. This review is an attempt to summarize new aspects of MC4R signalling in the context of its recently discovered alternative G protein coupling, and to give a perspective on how future research could improve our knowledge about the intertwining molecular mechanisms that are responsible for the regulation of energy homeostasis by the melanocortin system.
Collapse
Affiliation(s)
- Andreas Breit
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Goethestrasse 33, Ludwig-Maximilians-Universität München, 80336 München, Germany.
| | | | | | | | | | | |
Collapse
|
83
|
Oddi S, Dainese E, Fezza F, Lanuti M, Barcaroli D, De Laurenzi V, Centonze D, Maccarrone M. Functional characterization of putative cholesterol binding sequence (CRAC) in human type-1 cannabinoid receptor. J Neurochem 2011; 116:858-65. [PMID: 21214565 DOI: 10.1111/j.1471-4159.2010.07041.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Endocannabinoid signaling modulates a variety of neuroinflammatory and neurodegenerative diseases, mainly through the activation of type-1 and type-2 (CB(1)R and CB(2)R) cannabinoid receptors. CB(1)R is negatively regulated by membrane cholesterol, while CB(2)R is unaffected. Here, we identified in the transmembrane helix 7 of human CBRs a consensus sequence already known in other proteins as cholesterol recognition/interaction amino acid sequence and consensus pattern. As this motif is different in the two CBR subtypes, we mutated lysine 402 of CB(1)R into glycine, to obtain a cholesterol recognition/interaction amino acid sequence and consensus similar to that of CB(2)R. Both mutated and wild-type receptors were transiently expressed in human neuronal SH-SY5Y cells, and their localization and functioning were investigated using biochemical assays and immunofluorescence labelling. We found a reduced propensity of the mutant CB(1)R to reside in cholesterol-rich microdomains and, by means of fluorescence recovery after photobleaching analysis, we documented its loss of sensitivity to increased membrane cholesterol content. These results seem to uncover the existence of a new structural determinant in cannabinoid receptors, that is likely implicated in directing their interaction with cholesterol-rich microdomains of cell membranes.
Collapse
Affiliation(s)
- Sergio Oddi
- Department of Biomedical Sciences, University of Teramo, Teramo, Italy
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Depry C, Allen MD, Zhang J. Visualization of PKA activity in plasma membrane microdomains. ACTA ACUST UNITED AC 2011; 7:52-8. [DOI: 10.1039/c0mb00079e] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
85
|
Albizu L, Moreno JL, González-Maeso J, Sealfon SC. Heteromerization of G protein-coupled receptors: relevance to neurological disorders and neurotherapeutics. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2010; 9:636-50. [PMID: 20632964 PMCID: PMC3066024 DOI: 10.2174/187152710793361586] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 03/30/2010] [Indexed: 11/22/2022]
Abstract
Because G protein-coupled receptors (GPCRs) are numerous, widely expressed and involved in major physiological responses, they represent a relevant therapeutic target for drug discovery, particularly regarding pharmacological treatments of neurological disorders. Among the biological phenomena regulating receptor function, GPCR heteromerization is an important emerging area of interest and investigation. There is increasing evidence showing that heteromerization contributes to the pharmacological heterogeneity of GPCRs by modulating receptor ontogeny, activation and recycling. Although in many cases the physiological relevance of receptor heteromerization has not been fully established, the unique pharmacological and functional properties of heteromers are likely to lead to new strategies in clinical medicine. This review describes the main GPCR heteromers and their implications for major neurological disorders such as Parkinson's disease, schizophrenia and addiction. A better understanding of molecular mechanisms underlying drug interactions related to the targeting of receptor heteromers could provide more specific and efficient therapeutic agents for the treatment of brain diseases.
Collapse
Affiliation(s)
- Laura Albizu
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029, USA
- Center for Translational Systems Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - José L. Moreno
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Javier González-Maeso
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Stuart C. Sealfon
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029, USA
- Center for Translational Systems Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
86
|
Tashkin DP, Fabbri LM. Long-acting beta-agonists in the management of chronic obstructive pulmonary disease: current and future agents. Respir Res 2010; 11:149. [PMID: 21034447 PMCID: PMC2991288 DOI: 10.1186/1465-9921-11-149] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 10/29/2010] [Indexed: 02/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive airflow limitation and debilitating symptoms. For patients with moderate-to-severe COPD, long-acting bronchodilators are the mainstay of therapy; as symptoms progress, guidelines recommend combining bronchodilators from different classes to improve efficacy. Inhaled long-acting β2-agonists (LABAs) have been licensed for the treatment of COPD since the late 1990s and include formoterol and salmeterol. They improve lung function, symptoms of breathlessness and exercise limitation, health-related quality of life, and may reduce the rate of exacerbations, although not all patients achieve clinically meaningful improvements in symptoms or health related quality of life. In addition, LABAs have an acceptable safety profile, and are not associated with an increased risk of respiratory mortality, although adverse effects such as palpitations and tremor may limit the dose that can be tolerated. Formoterol and salmeterol have 12-hour durations of action; however, sustained bronchodilation is desirable in COPD. A LABA with a 24-hour duration of action could provide improvements in efficacy, compared with twice-daily LABAs, and the once-daily dosing regimen could help improve compliance. It is also desirable that a new LABA should demonstrate fast onset of action, and a safety profile at least comparable to existing LABAs.A number of novel LABAs with once-daily profiles are in development which may be judged against these criteria. Indacaterol, a LABA with a 24-hour duration of bronchodilation and fast onset of action, is the most advanced of these. Preliminary results from large clinical trials suggest indacaterol improves lung function compared with placebo and other long-acting bronchodilators. Other LABAs with a 24-hour duration of bronchodilation include carmoterol, vilanterol trifenatate and oldaterol, with early results indicating potential for once-daily dosing in humans.The introduction of once-daily LABAs also provides the opportunity to develop combination inhalers of two or more classes of once-daily long-acting bronchodilators, which may be advantageous for COPD patients through simplification of treatment regimens as well as improvements in efficacy. Once-daily LABAs used both alone and in combination with long-acting muscarinic antagonists represent a promising advance in the treatment of COPD, and are likely to further improve outcomes for patients.
Collapse
Affiliation(s)
- Donald P Tashkin
- David Geffen School of Medicine, Division of Pulmonary and Critical Care Medicine, UCLA, Los Angeles, California, USA
| | - Leonardo M Fabbri
- Department of Respiratory Diseases, University of Modena & Reggio Emilia, Via del Pozzo 71, I-41124 Modena, Italy
| |
Collapse
|
87
|
Paila YD, Jindal E, Goswami SK, Chattopadhyay A. Cholesterol depletion enhances adrenergic signaling in cardiac myocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1808:461-5. [PMID: 20851100 DOI: 10.1016/j.bbamem.2010.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 09/08/2010] [Accepted: 09/08/2010] [Indexed: 10/19/2022]
Abstract
Cardiac myocytes endogenously express α and β adrenergic receptors, prototypes of the G-protein coupled receptor superfamily. Depending upon the dose of norepinephrine (agonist) exposure, hypertrophy and apoptosis are initiated by differential induction of two discrete constituents of the transcription factor AP-1, i.e., FosB and Fra-1. We explored differential adrenergic signaling as a paradigm for understanding how cholesterol dictates cells to choose hypertrophy or apoptosis. For this, we used fosB and fra-1 promoter-reporter constructs for monitoring adrenergic signaling. We show that cholesterol depletion enhances norepinephrine-mediated signaling in cardiac myocytes. Importantly, this increased signaling is reduced to original level upon cholesterol replenishment. We used specific ligands for α and β adrenergic receptors and show that the enhanced signaling upon cholesterol depletion is a combined effect of both α and β adrenergic receptors. These results constitute the first report demonstrating the effect of cholesterol on adrenergic signaling using a direct end-point gene expression.
Collapse
Affiliation(s)
- Yamuna Devi Paila
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research Uppal Road, Hyderabad 500 007, India
| | | | | | | |
Collapse
|
88
|
Norman E, Cutler RG, Flannery R, Wang Y, Mattson MP. Plasma membrane sphingomyelin hydrolysis increases hippocampal neuron excitability by sphingosine-1-phosphate mediated mechanisms. J Neurochem 2010; 114:430-9. [PMID: 20456020 PMCID: PMC2910150 DOI: 10.1111/j.1471-4159.2010.06779.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Proteins that control the excitability of neurons, including voltage-dependent ion channels and neurotransmitter receptors, reside in a membrane lipid environment that includes sphingomyelin, but the influence of the metabolism of this lipid on excitability is unknown. Sphingomyelin in the plasma membrane can be cleaved by neutral sphingomyelinases (nSMase) to generate ceramides and sphingosine-1-phosphate (S1P) which have been shown to play a variety of roles in cellular signaling processes. We found that application of nSMase to hippocampal slices results in a selective enhancement in the population spike amplitude, resulting in fEPSP-PS potentiation of the CA3-CA1 schaeffer collateral synapse. Single cell recordings showed that nSMase activity increases action potential frequency in CA1 neurons in a reversible manner. Additional current clamp recordings showed that nSMase reduces the slow after-hyperpolarization after a burst of action potentials. Mass spectrometry-based measurements demonstrated that nSMase activity induces a rapid increase in the levels of ceramides and S1P in cells in hippocampal slices. The ability of nSMase to increase CA1 neuron excitability was blocked by an inhibitor of sphingosine kinase, the enzyme that converts ceramide to S1P. Moreover, direct intracellular application of S1P to CA1 neurons increased action potential firing. Our findings suggest roles for sphingomyelin metabolism and S1P in the positive regulation of the excitability of hippocampal neurons.
Collapse
Affiliation(s)
- Eric Norman
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH Biomedical Research Center, Baltimore, Maryland 21224, USA
| | | | | | | | | |
Collapse
|
89
|
Abstract
We performed molecular dynamics (MD) simulations of hydrated bilayers containing mixtures of dimyristoylphosphatidylcholine (DMPC) and cholesterol at various ratios, to study the effect of cholesterol concentration on its orientation, and to characterize the link between cholesterol tilt and overall phospholipid membrane organization. The simulations show a substantial probability for cholesterol molecules to transiently orient perpendicular to the bilayer normal, and suggest that cholesterol tilt may be an important factor for inducing membrane ordering. In particular, we find that as cholesterol concentration increases (1-40% cholesterol) the average cholesterol orientation changes in a manner strongly (anti)correlated with the variation in membrane thickness. Furthermore, cholesterol orientation is found to be determined by the aligning force exerted by other cholesterol molecules. To quantify this aligning field, we analyzed cholesterol orientation using, to our knowledge, the first estimates of the cholesterol tilt modulus chi from MD simulations. Our calculations suggest that the aligning field that determines chi is indeed strongly linked to sterol composition. This empirical parameter (chi) should therefore become a useful quantitative measure to describe cholesterol interaction with other lipids in bilayers, particularly in various coarse-grained force fields.
Collapse
Affiliation(s)
- George Khelashvili
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, NY, USA
| | - Georg Pabst
- Institute of Biophysics and Nanosystems Research, Austrian Academy of Sciences, A-8042 Graz, Austria
| | - Daniel Harries
- Institute of Chemistry and the Fritz Haber Research Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
90
|
Öner ŞS, Kaya Aİ, Onaran HO, Özcan G, Uğur Ö. β2-Adrenoceptor, Gs and adenylate cyclase coupling in purified detergent-resistant, low density membrane fractions. Eur J Pharmacol 2010; 630:42-52. [DOI: 10.1016/j.ejphar.2009.12.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 11/17/2009] [Accepted: 12/15/2009] [Indexed: 01/18/2023]
|
91
|
Soriano-Ursúa MA, Trujillo-Ferrara JG, Correa-Basurto J. Scope and difficulty in generating theoretical insights regarding ligand recognition and activation of the beta 2 adrenergic receptor. J Med Chem 2010; 53:923-932. [PMID: 19803522 DOI: 10.1021/jm900419a] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Marvin A Soriano-Ursúa
- Departamento de Bioquímica, Escuela Superior de Medicina, Instituto Politécnico Nacional,Mexico City, Mexico.
| | | | | |
Collapse
|
92
|
Vayttaden SJ, Friedman J, Tran TM, Rich TC, Dessauer CW, Clark RB. Quantitative modeling of GRK-mediated beta2AR regulation. PLoS Comput Biol 2010; 6:e1000647. [PMID: 20098494 PMCID: PMC2798957 DOI: 10.1371/journal.pcbi.1000647] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 12/15/2009] [Indexed: 11/18/2022] Open
Abstract
We developed a unified model of the GRK-mediated β2 adrenergic receptor (β2AR) regulation that simultaneously accounts for six different biochemical measurements of the system obtained over a wide range of agonist concentrations. Using a single deterministic model we accounted for (1) GRK phosphorylation in response to various full and partial agonists; (2) dephosphorylation of the GRK site on the β2AR; (3) β2AR internalization; (4) recycling of the β2AR post isoproterenol treatment; (5) β2AR desensitization; and (6) β2AR resensitization. Simulations of our model show that plasma membrane dephosphorylation and recycling of the phosphorylated receptor are necessary to adequately account for the measured dephosphorylation kinetics. We further used the model to predict the consequences of (1) modifying rates such as GRK phosphorylation of the receptor, arrestin binding and dissociation from the receptor, and receptor dephosphorylation that should reflect effects of knockdowns and overexpressions of these components; and (2) varying concentration and frequency of agonist stimulation “seen” by the β2AR to better mimic hormonal, neurophysiological and pharmacological stimulations of the β2AR. Exploring the consequences of rapid pulsatile agonist stimulation, we found that although resensitization was rapid, the β2AR system retained the memory of the previous stimuli and desensitized faster and much more strongly in response to subsequent stimuli. The latent memory that we predict is due to slower membrane dephosphorylation, which allows for progressive accumulation of phosphorylated receptor on the surface. This primes the receptor for faster arrestin binding on subsequent agonist activation leading to a greater extent of desensitization. In summary, the model is unique in accounting for the behavior of the β2AR system across multiple types of biochemical measurements using a single set of experimentally constrained parameters. It also provides insight into how the signaling machinery can retain memory of prior stimulation long after near complete resensitization has been achieved. The β2 adrenergic receptor (β2AR) is involved in regulating many cellular processes such as smooth muscle relaxation in the airways and the vasculature. Drugs that activate the β2AR are used in treating asthma and chronic obstructive pulmonary disorder (COPD), and prolonged use of these drugs leads to the loss of their effects. Thus, a dynamic model of how the β2AR responds to different drugs is fundamental to their rational use. In this study a consensus model of G protein coupled receptor kinase (GRK)-mediated receptor regulation was formulated based on quantitative measures of six processes involved in β2AR regulation. This model was then used to simulate the consequences of manipulating key rates associated with the GRK-mediated β2AR regulation, leading to predictions which will provide a useful framework for further tests and elaborations of the model in basic and clinical research.
Collapse
Affiliation(s)
- Sharat J Vayttaden
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States of America
| | | | | | | | | | | |
Collapse
|
93
|
Maccarrone M. Membrane environment and endocannabinoid signaling. Front Physiol 2010; 1:140. [PMID: 21423380 PMCID: PMC3059985 DOI: 10.3389/fphys.2010.00140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 10/06/2010] [Indexed: 01/01/2023] Open
Affiliation(s)
- Mauro Maccarrone
- Department of Biomedical Sciences, University of Teramo Teramo, Italy.
| |
Collapse
|
94
|
Terpager M, Scholl DJ, Kubale V, Martini L, Elling CE, Schwartz TW. Construction of covalently coupled, concatameric dimers of 7TM receptors. J Recept Signal Transduct Res 2009; 29:235-45. [PMID: 19747085 DOI: 10.1080/10799890903154217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
7TM receptors are easily fused to proteins such as G proteins and arrestin but because of the fact that their terminals are found on each side of the membrane they cannot be joined directly in covalent dimers. Here, we use an artificial connector comprising a transmembrane helix composed of Leu-Ala repeats flanked by flexible spacers and positively charged residues to ensure correct inside-out orientation plus an extracellular HA-tag to construct covalently coupled dimers of 7TM receptors. Such 15 TM concatameric homo- and heterodimers of the beta(2)-adrenergic and the NK(1) receptors, which normally do not dimerize with each other, were expressed surprisingly well at the cell surface, where they bound ligands and activated signal transduction in a manner rather similar to the corresponding wild-type receptors. The concatameric heterodimers internalized upon stimulation with agonists for either of the protomers, which was not observed upon simple coexpression of the two receptors. It is concluded that covalently joined 7TM receptor dimers with surprisingly normal receptor properties can be constructed with use of an artificial transmembrane connector, which perhaps can be used to fuse other membrane proteins.
Collapse
Affiliation(s)
- Marie Terpager
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
95
|
Abulrob A, Lu Z, Baumann E, Vobornik D, Taylor R, Stanimirovic D, Johnston LJ. Nanoscale imaging of epidermal growth factor receptor clustering: effects of inhibitors. J Biol Chem 2009; 285:3145-56. [PMID: 19959837 DOI: 10.1074/jbc.m109.073338] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The development of some solid tumors is associated with overexpression of the epidermal growth factor receptor (EGFR) and often correlates with poor prognosis. Near field scanning optical microscopy, a technique with subdiffraction-limited optical resolution, was used to examine the influence of two inhibitors (the chimeric 225 antibody and tyrosine phosphorylation inhibitor AG1478) on the nanoscale clustering of EGFR in HeLa cells. The EGFR is organized in small clusters, average diameter of 150 nm, on the plasma membrane for both control and EGF-treated cells. The numbers of receptors in individual clusters vary from as few as one or two proteins to greater than 100. Both inhibitors yield an increased cluster density and an increase in the fraction of clusters with smaller diameters and fewer receptors. Exposure to AG1478 also decreases the fraction of EGFR that colocalizes with both rafts and caveolae. EGF stimulation results in a significant loss of the full-length EGFR from the plasma membrane with the concomitant appearance of low molecular mass proteolytic products. By contrast, AG1478 reduces the level of EGFR degradation. Changes in receptor clustering provide one mechanism for regulating EGFR signaling and are relevant to the design of strategies for therapeutic interventions based on modulating EGFR signaling.
Collapse
Affiliation(s)
- Abedelnasser Abulrob
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario K1A 0R5, Canada.
| | | | | | | | | | | | | |
Collapse
|
96
|
Lipid membrane interactions of indacaterol and salmeterol: Do they influence their pharmacological properties? Eur J Pharm Sci 2009; 38:533-47. [DOI: 10.1016/j.ejps.2009.10.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 09/28/2009] [Accepted: 10/01/2009] [Indexed: 01/24/2023]
|
97
|
Allen JA, Yu JZ, Dave RH, Bhatnagar A, Roth BL, Rasenick MM. Caveolin-1 and lipid microdomains regulate Gs trafficking and attenuate Gs/adenylyl cyclase signaling. Mol Pharmacol 2009; 76:1082-93. [PMID: 19696145 PMCID: PMC2774991 DOI: 10.1124/mol.109.060160] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 08/18/2009] [Indexed: 01/06/2023] Open
Abstract
Lipid rafts and caveolae are specialized membrane microdomains implicated in regulating G protein-coupled receptor signaling cascades. Previous studies have suggested that rafts/caveolae may regulate beta-adrenergic receptor/Galpha(s) signaling, but underlying molecular mechanisms are largely undefined. Using a simplified model system in C6 glioma cells, this study disrupts rafts/caveolae using both pharmacological and genetic approaches to test whether caveolin-1 and lipid microdomains regulate G(s) trafficking and signaling. Lipid rafts/caveolae were disrupted in C6 cells by either short-term cholesterol chelation using methyl-beta-cyclodextrin or by stable knockdown of caveolin-1 and -2 by RNA interference. In imaging studies examining Galpha(s)-GFP during signaling, stimulation with the betaAR agonist isoproterenol resulted in internalization of Galpha(s)-GFP; however, this trafficking was blocked by methyl-beta-cyclodextrin or by caveolin knockdown. Caveolin knockdown significantly decreased Galpha(s) localization in detergent insoluble lipid raft/caveolae membrane fractions, suggesting that caveolin localizes a portion of Galpha(s) to these membrane microdomains. Methyl-beta-cyclodextrin or caveolin knockdown significantly increased isoproterenol or thyrotropin-stimulated cAMP accumulation. Furthermore, forskolin- and aluminum tetrafluoride-stimulated adenylyl cyclase activity was significantly increased by caveolin knockdown in cells or in brain membranes obtained from caveolin-1 knockout mice, indicating that caveolin attenuates signaling at the level of Galpha(s)/adenylyl cyclase and distal to GPCRs. Taken together, these results demonstrate that caveolin-1 and lipid microdomains exert a major effect on Galpha(s) trafficking and signaling. It is suggested that lipid rafts/caveolae are sites that remove Galpha(s) from membrane signaling cascades and caveolins might dampen globally Galpha(s)/adenylyl cyclase/cAMP signaling.
Collapse
Affiliation(s)
- John A Allen
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago (UIC), Chicago, IL 60612-7342, USA
| | | | | | | | | | | |
Collapse
|
98
|
Fallahi-Sichani M, Linderman JJ. Lipid raft-mediated regulation of G-protein coupled receptor signaling by ligands which influence receptor dimerization: a computational study. PLoS One 2009; 4:e6604. [PMID: 19668374 PMCID: PMC2719103 DOI: 10.1371/journal.pone.0006604] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 07/22/2009] [Indexed: 11/19/2022] Open
Abstract
G-protein coupled receptors (GPCRs) are the largest family of cell surface receptors; they activate heterotrimeric G-proteins in response to ligand stimulation. Although many GPCRs have been shown to form homo- and/or heterodimers on the cell membrane, the purpose of this dimerization is not known. Recent research has shown that receptor dimerization may have a role in organization of receptors on the cell surface. In addition, microdomains on the cell membrane termed lipid rafts have been shown to play a role in GPCR localization. Using a combination of stochastic (Monte Carlo) and deterministic modeling, we propose a novel mechanism for lipid raft partitioning of GPCRs based on reversible dimerization of receptors and then demonstrate that such localization can affect GPCR signaling. Modeling results are consistent with a variety of experimental data indicating that lipid rafts have a role in amplification or attenuation of G-protein signaling. Thus our work suggests a new mechanism by which dimerization-inducing or inhibiting characteristics of ligands can influence GPCR signaling by controlling receptor organization on the cell membrane.
Collapse
Affiliation(s)
- Mohammad Fallahi-Sichani
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer J. Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
99
|
Wetzel G, Heine M, Rohwedder A, Naim HY. Impact of glycosylation and detergent-resistant membranes on the function of intestinal sucrase-isomaltase. Biol Chem 2009; 390:545-9. [DOI: 10.1515/bc.2009.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Sucrase-isomaltase (SI) is a highly N- and O-glycosylated intestinal brush border membrane protein. SI is sorted with high fidelity to the apical membrane via O-linked glycans that mediate its association with lipid rafts or detergent-resistant membranes (DRMs). Here, we show that N- and O-glycosylation and DRMs are implicated in the regulation of the function of SI in intestinal Caco-2 cells. The activities of sucrase (SUC) and isomaltase (IM) increase substantially in DRMs when N- and O-glycosylation patterns are intact. Disruption of DRMs by solubilization with Triton X-100 at 37°C substantially reduces the activities of SUC and IM. Furthermore, modulation of O-glycosylation by benzyl-2-acetamido-2-deoxy-α-d-galactopyranoside and N-glycosylation by deoxymannojirimycin is linked to a decreased capacity of SI to associate with DRMs, with a subsequent reduction of the enzymatic activities of SUC and IM. This is the first report that reveals a direct role of N- and O-glycans in association with DRMs in regulating the function of a membrane glycoprotein.
Collapse
|
100
|
Levitt ES, Clark MJ, Jenkins PM, Martens JR, Traynor JR. Differential effect of membrane cholesterol removal on mu- and delta-opioid receptors: a parallel comparison of acute and chronic signaling to adenylyl cyclase. J Biol Chem 2009; 284:22108-22122. [PMID: 19520863 DOI: 10.1074/jbc.m109.030411] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
According to the lipid raft theory, the plasma membrane contains small domains enriched in cholesterol and sphingolipid, which may serve as platforms to organize membrane proteins. Using methyl-beta-cyclodextrin (MbetaCD) to deplete membrane cholesterol, many G protein-coupled receptors have been shown to depend on putative lipid rafts for proper signaling. Here we examine the hypothesis that treatment of HEK293 cells stably expressing FLAG-tagged mu-opioid receptors (HEK FLAG-mu) or delta-opioid receptors (HEK FLAG-delta) with MbetaCD will reduce opioid receptor signaling to adenylyl cyclase. The ability of the mu-opioid agonist [D-Ala2,N-Me-Phe4,Gly5-ol]enkephalin to acutely inhibit adenylyl cyclase or to cause sensitization of adenylyl cyclase following chronic treatment was attenuated with MbetaCD. These effects were due to removal of cholesterol, because replenishment of cholesterol restored [D-Ala2,N-Me-Phe4,Gly5-ol]enkephalin responses back to control values, and were confirmed in SH-SY5Y cells endogenously expressing mu-opioid receptors. The effects of MbetaCD may be due to uncoupling of the mu receptor from G proteins but were not because of decreases in receptor number and were not mimicked by cytoskeleton disruption. In contrast to the results in HEK FLAG-mu cells, MbetaCD treatment of HEK FLAG-delta cells had no effect on acute inhibition or sensitization of adenylyl cyclase by delta-opioid agonists. The differential responses of mu- and delta-opioid agonists to cholesterol depletion suggest that mu-opioid receptors are more dependent on cholesterol for efficient signaling than delta receptors and can be partly explained by localization of mu- but not delta-opioid receptors in cholesterol- and caveolin-enriched membrane domains.
Collapse
Affiliation(s)
| | - Mary J Clark
- Department of Pharmacology, Ann Arbor, Michigan 48109
| | | | | | - John R Traynor
- Department of Pharmacology, Ann Arbor, Michigan 48109; Substance Abuse Research Center, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|